current views on hiv-1 latency, persistence, and cure · et al. 2009; fukazawa et al. 2015; kim and...

15
Current views on HIV-1 latency, persistence, and cure Zora Melkova 1,2 & Prakash Shankaran 1 & Michaela Madlenakova 1,2 & Josef Bodor 2 Received: 10 May 2016 /Accepted: 20 September 2016 # Institute of Microbiology, Academy of Sciences of the Czech Republic, v.v.i. 2016 Abstract HIV-1 infection cannot be cured as it persists in latently infected cells that are targeted neither by the immune system nor by available therapeutic approaches. Consequently, a lifelong therapy suppressing only the actively replicating virus is necessary. The latent reservoir has been defined and characterized in various experimental models and in human patients, allowing research and development of approaches targeting individual steps critical for HIV-1 la- tency establishment, maintenance, and reactivation. However, additional mechanisms and processes driving the remaining low-level HIV-1 replication in the presence of the suppressive therapy still remain to be identified and targeted. Current ap- proaches toward HIV-1 cure involve namely attempts to reac- tivate and purge HIV latently infected cells (so-called Bshock and kill^ strategy), as well as approaches involving gene ther- apy and/or gene editing and stem cell transplantation aiming at generation of cells resistant to HIV-1. This review summarizes current views and concepts underlying different approaches aiming at functional or sterilizing cure of HIV-1 infection. Introduction HIV/AIDS infection can be successfully treated and con- trolled with the combined antiretroviral therapy (cART) affecting different steps of HIV-1 replication cycle. Thus, HIV-1 infection could be viewed as a chronic disease with a relatively long life expectancy. However, cART, which is still not available to all in need, cannot cure HIV infection due to the presence of latently infected reservoir cells. The latent proviral DNA cannot be recognized by the immune system nor targeted by cART. Consequently, a lifelong therapy is necessary, which is expensive and leads to various complica- tions and treatment failures. Therefore, new approaches to- ward functional or sterilizing cure are intensively explored namely attempts to reactivate and purge HIV-1 latently infect- ed cells (so-called Bshock and kill^ strategy), as well as ap- proaches involving gene therapy and/or gene editing and stem cell transplantation aiming at generation of cells resistant to HIV-1. The ongoing research focuses especially on the mech- anisms of establishment and maintenance of the latent reser- voir, assessment of its size and composition, as well as on stimulation of the innate and specific immunity to promote HIV-1 clearance. HIV-1 latency Retrovirus replication cycle is specific by a step of reverse transcription and a consequent stable integration of the provi- ral DNA into the host cell genome. Depending on the status of the host cell, either the HIV-1 provirus can be immediately expressed and the virus replication cycle can proceed further or the provirus can become dormant and wait until the latently infected cell encounters the right stimulus (often a specific, possibly rare antigen) and becomes activated. It is the very presence of the latently infected cells that makes HIV-1 infection incurable as these cells serve as a source of virus rebound after a discontinuation or failure of the antiretroviral therapy. After the activation of the host cell or specific * Zora Melkova [email protected] 1 Department of Immunology and Microbiology, 1st Faculty of Medicine, Charles University, Studnickova 7, 128 00 Prague 2, Czech Republic 2 BIOCEV, Biotechnology and Biomedicine Center of the Academy of Sciences and Charles University in Vestec, Průmyslová 595, 252 50 Vestec, Czech Republic Folia Microbiol DOI 10.1007/s12223-016-0474-7

Upload: others

Post on 05-Sep-2019

4 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Current views on HIV-1 latency, persistence, and cure · et al. 2009; Fukazawa et al. 2015; Kim and Siliciano 2016; Van Lint et al. 2013).Additionally,thelatentreservoirisread-ily

Current views on HIV-1 latency, persistence, and cure

Zora Melkova1,2 & Prakash Shankaran1& Michaela Madlenakova1,2 & Josef Bodor2

Received: 10 May 2016 /Accepted: 20 September 2016# Institute of Microbiology, Academy of Sciences of the Czech Republic, v.v.i. 2016

Abstract HIV-1 infection cannot be cured as it persists inlatently infected cells that are targeted neither by the immunesystem nor by available therapeutic approaches.Consequently, a lifelong therapy suppressing only the activelyreplicating virus is necessary. The latent reservoir has beendefined and characterized in various experimental modelsand in human patients, allowing research and developmentof approaches targeting individual steps critical for HIV-1 la-tency establishment, maintenance, and reactivation. However,additional mechanisms and processes driving the remaininglow-level HIV-1 replication in the presence of the suppressivetherapy still remain to be identified and targeted. Current ap-proaches toward HIV-1 cure involve namely attempts to reac-tivate and purge HIV latently infected cells (so-called Bshockand kill^ strategy), as well as approaches involving gene ther-apy and/or gene editing and stem cell transplantation aiming atgeneration of cells resistant to HIV-1. This review summarizescurrent views and concepts underlying different approachesaiming at functional or sterilizing cure of HIV-1 infection.

Introduction

HIV/AIDS infection can be successfully treated and con-trolled with the combined antiretroviral therapy (cART)

affecting different steps of HIV-1 replication cycle. Thus,HIV-1 infection could be viewed as a chronic disease with arelatively long life expectancy. However, cART, which is stillnot available to all in need, cannot cure HIV infection due tothe presence of latently infected reservoir cells. The latentproviral DNA cannot be recognized by the immune systemnor targeted by cART. Consequently, a lifelong therapy isnecessary, which is expensive and leads to various complica-tions and treatment failures. Therefore, new approaches to-ward functional or sterilizing cure are intensively explored –namely attempts to reactivate and purge HIV-1 latently infect-ed cells (so-called Bshock and kill^ strategy), as well as ap-proaches involving gene therapy and/or gene editing and stemcell transplantation aiming at generation of cells resistant toHIV-1. The ongoing research focuses especially on the mech-anisms of establishment and maintenance of the latent reser-voir, assessment of its size and composition, as well as onstimulation of the innate and specific immunity to promoteHIV-1 clearance.

HIV-1 latency

Retrovirus replication cycle is specific by a step of reversetranscription and a consequent stable integration of the provi-ral DNA into the host cell genome. Depending on the status ofthe host cell, either the HIV-1 provirus can be immediatelyexpressed and the virus replication cycle can proceed furtheror the provirus can become dormant and wait until the latentlyinfected cell encounters the right stimulus (often a specific,possibly rare antigen) and becomes activated. It is thevery presence of the latently infected cells that makes HIV-1infection incurable as these cells serve as a source of virusrebound after a discontinuation or failure of the antiretroviraltherapy. After the activation of the host cell or specific

* Zora [email protected]

1 Department of Immunology and Microbiology, 1st Faculty ofMedicine, Charles University, Studnickova 7, 128 00 Prague2, Czech Republic

2 BIOCEV, Biotechnology and Biomedicine Center of the Academy ofSciences and Charles University in Vestec, Průmyslová 595, 25250 Vestec, Czech Republic

Folia MicrobiolDOI 10.1007/s12223-016-0474-7

Page 2: Current views on HIV-1 latency, persistence, and cure · et al. 2009; Fukazawa et al. 2015; Kim and Siliciano 2016; Van Lint et al. 2013).Additionally,thelatentreservoirisread-ily

changes in the epigenetic regulation, the chromatin status andavailability of transcription and other factors change and HIV-1 replication can restart (Fig. 1).

HIV-1 persistence during therapy

HIV-1 infects namely CD4+ T lymphocytes but also myeloidcells like macrophages, microglia, astrocytes, and dendriticcells, even though to a lesser extent. Acute infection ofCD4+ T cells usually leads to cell death, but occasionally,these cells survive and revert back to a resting memory state.Alternatively, latency may be established directly in restingCD4+ T cells. Resting memory cells, including the latentlyinfected ones, then persist and/or are replenished by homeo-static proliferation. In contrast, macrophages are resistant tocytopathic effects of HIV-1 infection and support virus persis-tence in various anatomical sanctuaries as tissue resident mac-rophages (Kim and Siliciano 2016; Kumar et al. 2015;McKinstry et al. 2010).

After initiation of cART, plasma viremia and the level ofHIV-1-infected cells in peripheral blood decay with a wellcharacterized kinetics based on populations with a differentturnover contributing to plasma viremia (Hilldorfer et al.2012) (Fig. 2). A rapid decline of plasma viremia duringfirst two phases indicates the efficiency of antiretroviral

drugs. Phase I represents the turnover of the free virus(half-life from minutes to hours) and mainly of productivelyinfected CD4+ T cells (half-life of 1–2 days; Ho et al. 1995;Perelson et al. 1997; Perelson et al. 1996; Wei et al. 1995).Consequently, phase II corresponds to the decay of cellsmore resistant to HIV-induced cytopathic effect (partiallyactivated T cells and cells of the monocyte-macrophage lin-eage with half-life of 2–4 weeks; Perelson et al. 1997; Shanand Siliciano 2013). The following phase III with a very lowdecay kinetics (half-life of 273 days) proceeds after severalyears into phase IV with a remaining stable low level plasmaviremia that does not decrease any more (Hilldorfer et al.2012; Maldarelli et al. 2007). Despite the very low plasmaviremia below the limit of clinical assays (<50 copies/ml),which is detectable with only ultrasensitive assays (about 3copies/ml), cell-associated proviral DNA (prDNA) and RNA(caRNA) are commonly detected by PCR-based assays inperipheral blood mononuclear cells (PBMCs) during thisphase (Palmer et al. 2008; Palmer et al. 2003) (Fig. 2).The presence of the phase IV indicates the existence of ad-ditional cell populations that do not succumb to virus-induced cytopathic effects and/or either are refractory tocART (proviral DNA or transcription cannot be targeted bycART) or persist in anatomical sanctuaries that are notaccessed by cART. Reactivation of the latently infected cells,which both persist and are replenished by homeostatic

HDACs DNMTs HMTs

NF-κB, NFAT, Sp1HAT (CBP/p300)TAT + p-TEFb etc.

Ac�vely transcribed provirus – open chroma�n

NF-κBNFAT

Silenced provirus – repressed chroma�n

Produc�ve elonga�on

Reac�va�on of HIV provirus

NELF

7SK snRNP DSIF

P

P

HEXIM1 LARP7MePCE7SK snRNA

HIV LTR

P

Transcrip�on blocked

Transcrip�on ac�ve

p-TEFb

TAT

TAR RNA

HIV LTR

RNAPII

Fig. 1 Latency and reactivation(simplified scheme). HIV-1latency is characterized by arepressed chromatin, presence ofhistone deacetylases (HDACs),histone methyltransferases(HMTs), and DNA methylases(DMTs), as well as lack oftranscription factors, resulting in atranscription block. Reactivationis associated with epigeneticchanges that lead to openchromatin structure, namelypresence of histoneacetyltransferases (HATs),nuclear translocation oftranscription initiation factors NF-κB and NFAT, increased levels ofTat and formation of its complexwith p-TEFb. Tat-p-TEFbcomplex binds to TAR RNA,resolving promoter-proximalpausing of RNAP II and allowingefficient transcription elongation.p-TEFb can be sequestered in the7SK snRNP inhibitory complex

Folia Microbiol

Page 3: Current views on HIV-1 latency, persistence, and cure · et al. 2009; Fukazawa et al. 2015; Kim and Siliciano 2016; Van Lint et al. 2013).Additionally,thelatentreservoirisread-ily

proliferation (Chomont et al. 2009; Kim and Siliciano 2016),is viewed as the most relevant source of the new HIV RNA(Shan and Siliciano 2013). However, lower levels ofantiretrovirals (ARV) found in anatomical/immunologicalsanctuaries like brain, genital tract, gut mucosa, or lymphnodes can support the ongoing HIV replication (Fletcheret al. 2014; Huang et al. 2016; Massanella et al. 2016).Based on sequence determination of HIV-1 present in lymphnodes and blood at several time-points after initiation ofcART, the mathematical modeling of virus evolution andtrafficking between the two compartments with low and highlevels of ARV, respectively, supported the view that evolu-tion of drug-sensitive viruses in lymph nodes can keepreplenishing the viral reservoir despite the apparently effi-cient cART (Lorenzo-Redondo et al. 2016). Lately, it wasdescribed that SIV/HIV-1 may continue to replicate within Bcell follicles due to follicular exclusion of CD8+ T cells anddysregulated responses of follicular regulatory T cells (TFR)and follicular T helper cells (TFH; Fukazawa et al. 2015;Miles et al. 2015; Saison et al. 2014; Tran et al. 2008).Furthermore, in addition to latently infected long-lived mem-ory T cells and tissue resident macrophages (Avalos et al.2016; Gludish et al. 2015), an evidence for existence oftissue resident memory T cells (TRM) emerged (Farber2015; Farber et al. 2014).

Another cause of the residual low-level HIV replicationunder cART could involve the immune hyperactivation in-duced by various mechanisms. In addition to the residual pro-ductive infection that is likely to rise immune responses, thereis a more frequent (95 %) abortive infection of resting non-permissive CD4+ T cells. Due to the cytoplasmic DNA-sensing mechanisms like IFI-16, cGAS, and PQBP-1 (Gaoet al. 2013; Thompson et al. 2014; Yoh et al. 2015), these cellsmight die by pyroptosis, a very inflammatory type of pro-grammed cell death (Doitsh et al. 2014; Monroe et al. 2014).Furthermore, HIV-1 replication and disruption of the intestinal

barrier lead to microbial translocation, stimulation of innateimmune responses, and depletion of CD4 Th17 cells, a defectpersisting even after initiation of cART (Chege et al. 2011;Schuetz et al. 2014). Finally, the activation and dysregulatedfunction of regulatory T cells (Tregs), critical modulators ofimmune responses, apparently also contribute the immunehyperactivation (Mendez-Lagares et al. 2014; Saison et al.2014). The residual low-level HIV replication that leads toan elevated immune activation thus further stimulates HIVreplication, generating a vicious cycle.

In summary, HIV persistence during cART is supported byhomeostatic and antigen-induced proliferation of latently in-fected cells, ongoing replication in the sanctuaries as well asby increased immune activation and inflammation (Chomontet al. 2009; Fukazawa et al. 2015; Kim and Siliciano 2016;Van Lint et al. 2013). Additionally, the latent reservoir is read-ily being replenished during episodes of viremia due to atreatment failure or other conditions of incomplete or missingpharmaceutical control.

Models of HIV-1 latency

Our knowledge of the mechanisms, maintenance, and reacti-vation of the latent reservoir is based namely on variousin vitro and in vivo models as the presence of latently infectedcells in human body is very low, around 1–10 millions in thewhole organism, and difficult to study (Crooks et al. 2015;Finzi et al. 1999; Massanella and Richman 2016; Silicianoet al. 2003).

Individual models of HIV latency reproduce to some extentcertain aspects of the complex situation in vivo and allow forstudies of certain latently infected populations or specific as-pects of latently infected cells. Models used to study HIV-1latency in vitro include HIV-infected cell lines (immortalizedlymphocytic or monocytic cells like J-Lat, ACH-2, U1;

1.00E-07

1.00E-06

1.00E-05

1.00E-04

1.00E-03

1.00E-02

1.00E-01

1.00E+00

1.00E+01

1.00E-01

1.00E+00

1.00E+01

1.00E+02

1.00E+03

1.00E+04

1.00E+05

1.00E+06

1.00E+07

-1 0 1 2 3 4 5 6 7 8

HIV

DNA

in P

BMCs

Plas

ma

HIV

RNA

(cop

ies/

mL)

Years on ART

Plasma HIV RNA (copies/ml) HIV DNA in PBMCs

h\\\\\\\\\\\\\\\\\\\\\\\\\\`

Ini�ate ART

Phase It½ = 1.5 days

Phase IIt½ = 28 days

Phase IIIt½ = 273 days Phase IV

t½ = ∞

Fig. 2 Comparison of HIV-1RNA and DNA decay curvesupon introduction of cART. Afterinitiation of cART, plasmaviremia and the level of HIV-1infected cells in peripheral blooddecay with a well characterizedkinetics based on populationswith different turnoverscontributing to plasma viremia(Phase I–IV). HIV-1 DNA revealsa relatively smaller decrease.Adapted from Hilldorfer et al.(2012) and Margolis et al. (2016)

Folia Microbiol

Page 4: Current views on HIV-1 latency, persistence, and cure · et al. 2009; Fukazawa et al. 2015; Kim and Siliciano 2016; Van Lint et al. 2013).Additionally,thelatentreservoirisread-ily

Clouse et al. 1989; Folks et al. 1989; Folks et al. 1987; Jordanet al. 2003), primary cell models (derived from HIV-1 nega-tive donor PBMCs by infection with a particular HIV-1 isolateor a recombinant), and resting CD4 cells (derived fromHIV-1-infected patients; Spina et al. 2013). While the cell lines arecommonly available and easy to handle, they reveal severalaspects that often make them behave differently from the sit-uation in vivo (namely, clonality based on the integration siteor mutations of specific HIV-1 or host cell sequences).

Primary cell models might seem to be closer to the situationin vivo, while the percentage of latently infected cells avail-able for further studies is much higher than in vivo. Differentprimary cell models developed by different laboratories useeither resting or activated CD4+ T cells that are infected withwild-type (typically NL4-3) or recombinant HIV-1 (often ex-pressing a fluorescent reporter gene like EGFP) and treatedwith combinations of several cytokines or chemokines. Afterestablishment of the infection, latency is induced by ARVsfollowed by reactivation by different means. The individualmodels differ in the target cell population, percentage of la-tently infected cells generated, type and time of readout(Bosque and Planelles 2009; Gondois-Rey et al. 2006;Lassen et al. 2012; Saleh et al. 2007; Spina et al. 2013;Tyagi et al. 2010; Yang et al. 2009b). Nevertheless, it is ques-tionable how closely these models reflect the situation in vivo,and how much they are biased due to the experimental setupand mode of readout.

Resting CD4 cells derived from HIV-1-infected patientscultured and stimulated ex vivo thus seem to be the mostrelevant in vitro model. However, their use is limited by a verylow presence of latently infected cells, high background rateof defective integrated proviruses, and difficulties of any anal-ysis due to a very high background of uninfected cells (prob-lems with sensitivity and specificity).

The in vivo models of HIV latency include namely differ-ent types of humanized mice and macaques infected with HIV,SIV, or various recombinants. The advantage of humanizedmice consists in their relative affordability and ease of han-dling; on the other hand, their preparation is tedious whilegraft-versus-host disease (GvHD) and other differences,namely due to their genetic background, the way of immunereconstitution with human tissues, and lineage precursors,may limit their use and relevance of the results.

Humanized mice used for HIV research were originallybased on SCID mice (severe combined immunodeficiencymice) that were irradiated and then transplanted with fetalhuman thymus and liver (SCID-hu Thy/Liv). In this model,latent infection is established during thymopoiesis (deactiva-tion phase), leading to generation of latently infected naïveCD4+ T cells. Among other limitations, this model does notprovide an efficient peripheral reconstitution and human cellsare found in relatively small numbers (Brooks et al. 2001;Marsden et al. 2012).

Lately, BLT mice (bone marrow-liver-thymus humanizedmice) are considered as a better model for complex studies ofHIV reservoirs and latency reversing agents (LRA) as theyprovide a robust peripheral reconstitution. These mice aremost commonly based on irradiated NSG mice (NOD/SCID-gamma chain null mice) transplanted with fetal humanthymus and liver and then reconstituted with bone marrow orpurified CD34+ stem cells (Donahue and Wainberg 2013).This particular combination results in high-level systemic re-constitution of all human leukocyte lineages with improved Tcell maturation and selection in a thymic environment and ingeneration of latently infected naïve and resting CD4+ T cells(Denton et al. 2012). There are also modifications of thesemice using reconstitution with only discrete cell types like Tcells or macrophages (T cell-only mice (ToM); myeloid-onlymice (MoM); Honeycutt et al. 2013; Honeycutt et al. 2016),allowing studies of the role of the individual cell types in theestablishment of latent reservoir or reactivation.

Although the use of BLT mouse model is valuable for theHIV studies, its important limitation consists in the develop-ment of GvHD, typically around 6 months after engraftment(Karpel et al. 2015). Further, these mice are unable to developproper HIV-specific adaptive immune responses consisting inhigh levels of hyper-mutated, class-switched IgG antibodiesas human cells of non-hematopoietic origin, namely thosegiving rise to stromal cells, are not transplanted, and second-ary lymphoid organs do not properly develop (Malhotra et al.2013; Wang et al. 2011).

Yet another BLT model, based on human Rag2−/−γc −/−

mice (Choudhary et al. 2012; Traggiai et al. 2004; Zhanget al. 2007), was recently developed and led to identificationof central memory CD4+ T cells (TCM) as the main latentlyinfected population after suppressive cART similarly as inhuman (Choudhary et al. 2012; Donahue and Wainberg2013). This model was further improved by generation ofhumanized TKO-BLT mice (triple knockout-BLT mice) on aC57Bl/6 background (C57BL/6 Rag2−/−γc−/−CD47−/−) inwhich the GvDH was much reduced, complement was func-tional, and secondary lymphoid organs with a well organizedarchitecture and virus-specific immune responses were devel-oped (Lavender et al. 2013).

A model closest to HIV-1 infection in human is infection ofnon-human primates (NHP), namely rhesus macaques, withvarious SIV strains. It is believed that different SIVs crossedthe species barrier into humans many times, namely SIVsmmnaturally infecting sooty mangabeys and SIVcpz infectingchimpanzees resulted in HIV-2 and certain clades of HIV-1(Hirsch et al. 1989; Huet et al. 1990; Sharp and Hahn 2011).

The NHP models reveal many important features compa-rable to HIV-1-infected humans like anatomy, physiology, im-mune system, infectious agent itself, and susceptibility to an-tiretroviral treatment (Gardner and Luciw 2008; Policicchioet al. 2016). The use of NHP models has been essential for

Folia Microbiol

Page 5: Current views on HIV-1 latency, persistence, and cure · et al. 2009; Fukazawa et al. 2015; Kim and Siliciano 2016; Van Lint et al. 2013).Additionally,thelatentreservoirisread-ily

understanding pathogenesis of HIV/AIDS as well as for stud-ies of different therapeutic and vaccination approaches. Manyconclusions could be also inferred by comparing immune re-sponses to SIV infection in the natural and unnatural hosts,sooty mangabeys and rhesus macaques, respectively, in whichthe SIV infection results in non-pathogenic or pathogenic con-ditions. There is a high viremia in both hosts but a very littleloss of CD4+ T cells and preservation of architecture andfunction of lymph nodes in the natural host. In the gut of thenatural host, CD4+ T cells are only moderately depleted andnumbers and functions of Th17 cells remain preserved, whilemicrobial translocation is lacking. Further, TFH and TCM areonly weakly infected (Ploquin et al. 2016).

Additionally, recombinant viruses like RT-SHIV or SHIVcontaining different parts of HIV-1 genome can be used toovercome certain differences between HIV-1 and SIVs, e.g.,in studies involving inhibitors specific for HIV reverse tran-scriptase (Jiang et al. 2009; Ndung’u et al. 2001). One impor-tant difference in comparison with HIV-1 is that most SIVs(and HIV-2) encode a vpx protein that allows virus infectionalso in non-dividing cells as it targets for degradation cellularSAMHD-1, an enzyme that would hydrolyze dNTPs and thusdecrease reverse transcription (Hofmann et al. 2012). Despitemany advantages, a major drawback of using macaques con-sists in their cost and ethical and legal regulations of their use.

Obviously, the most relevant approach to study HIV laten-cy and reactivation is in HIV-infected human patients in clin-ical studies. However, performance of clinical studies is strict-ly regulated, expensive, and must be preceded by extensivepre-clinical testing (including animal models). Additionally,human studies never provide really homogeneous and repro-ducible experimental settings with all necessary controls.

In summary, the results obtained in various in vitro andin vivo models and in human patients indicate that the latentHIV-1 reservoir is represented mainly by latently infectedresting CD4+ Tcells, long-lived central memory CD4+ Tcells(TCM cells), and transitional memory CD4+ T cells (TTM

cells). In addition to these cells, latent reservoir may comprisealso other cell populations like CD34+ hematopoietic progen-itor cells, naïve CD4+ T cells, CD4+ memory stem cells(TSCM cells) or γδ T cells, as well as myeloid cells likemacrophages and dendritic cells (Buzon et al. 2014; Carteret al. 2010; Chomont et al. 2009; Honeycutt et al. 2016;Soriano-Sarabia et al. 2015; Wightman et al. 2010). The im-portance of these cells constituting the latent reservoir consistsin their different survivals and stabilities as well as in differentrequirements for signaling and activation and thus HIV-1 pro-virus induction (Archin et al. 2014b).

Assessment of the size of the latent reservoir

Under suppressive cART, plasma viremia (virion-associatedunspliced RNA) is undetectable or below the detection limit ofcommon commercial assays (<50 copies/ml). However,markers of immune hyperactivation persist and cell-associated HIV RNA can be readily detected both in periph-eral blood and tissues like lymph nodes, tonsils, gut, or testes.Thus, in order to determine the effect of treatment intensifica-tion strategies and namely efficiency of LRA explored fortherapeutic reactivation, availability of specific and sensitivemethods allowing an accurate assessment of the size of thelatent reservoir is crucial (Fig. 3).

The latently infected cells can be defined as cells harboringquiescent, replication-competent provirus. A gold standard indetermination of latently infected cells is a quantitative virusoutgrowth assay (qVOA) that is very material-, labor-, andcost-demanding. It requires large volumes of patients’ bloodto isolate sufficient numbers of highly purified latently infect-ed resting CD4+ T cells and plate them in serial dilutionsalong with activated donor PBMCs (CD8 negative) or witha MOLT4/CCR5 cell line. It requires a 2–3-week incubationwith changes of medium and other additives (Finzi et al. 1997;Massanella and Richman 2016; Siliciano and Siliciano 2005).

Isola�on of RNA

Seminested 2-step RT-qPCR HIV msRNA

HIV msRNA

Isola�on of DNA

Seminested 2-step qPCRHIV prDNA

HIV prDNA

Quan�ta�on of HIV-1 DNA

Quan�ta�on of HIV-1 RNA

Limi�ng dillu�on assay+ cell lysis

Nested 1-step RT-PCRHIV msRNA

Frequency of cells withHIV msRNA

CD4+(PBMCs)

CD4+(PBMCs)

CD4+(PBMCs)

Fig. 3 PCR-based detection ofHIV-1 RNA and DNA.Determination of cell-associatedmultiply spliced RNA (msRNA)and proviral DNA (prDNA) usingtwo-step seminested (RT-) qPCRor using a combination of alimiting dilution assay and one-step nested RT-PCR (e.g.,TILDA; Bullen et al. 2014;Kiselinova et al. 2014; Procopioet al. 2015)

Folia Microbiol

Page 6: Current views on HIV-1 latency, persistence, and cure · et al. 2009; Fukazawa et al. 2015; Kim and Siliciano 2016; Van Lint et al. 2013).Additionally,thelatentreservoirisread-ily

However, this assay apparently underestimates the size of thelatent pool, as it has been already demonstrated that consecu-tive rounds of stimulation/reactivation of the isolated cellscould lead to increased proportion of the reactivated provi-ruses (Bruner et al. 2015; Ho et al. 2013). Further, only 1–2% of the body’s lymphocytes are present in peripheral blood,while most of them are found in tissues and the distribution oflatently infected cells might not be proportional (Blum andPabst 2007; Massanella and Richman 2016). The real size ofthe latent reservoir thus might be at least 60× higher thanobserved in a common qVOA, complicating the effort to cureHIV using latency reversal (Bruner et al. 2015; Ho et al.2013). Importantly, about 10 % of HIV-1 proviruses that re-main silent after the maximal stimulation were found to befully replication competent, suggesting that they might bereactivated in vivo upon different conditions than those usedex vivo during qVOA. Also, the induction of intact proviruseswas proposed to be stochastic, dependent on the levels of Tat,but independent of the cellular activation status (Ho et al.2013; Weinberger et al. 2005; Weinberger et al. 2008).

An analog of qVOA, mouse virus outgrowth assay(MVOA), allows to determine virus outgrowth in humanizedmice. It has been recently described to detect latently infectedcells with higher sensitivity than the standard qVOA as a largenumber of cells can be used and a GvHD promotes HIV-1reactivation. Nevertheless, it provides only qualitative results(Metcalf Pate et al. 2015).

At the other side is a PCR-based determination of the cell-associated DNA, which apparently overestimates the size ofthe latent pool, as most of the integrated proviruses are mutat-ed or incapable of reactivation for unknown reasons (Ho et al.2013; Sanchez et al. 1997). Depending on the primer/probesequences, total, integrated, or 2-long terminal repeat (2-LTR)circular DNA can be commonly detected (Murray et al. 2014;Pasternak et al. 2013). Finally, determination of cell-associated RNA (caRNA) using various combinations ofPCR-based techniques like seminested RT-qPCR or ddPCR(Bullen et al. 2014; Kiselinova et al. 2014) seems to provide amore relevant marker of viral persistence and/or reactivation.However, it is important to use approaches distinguishing be-tween commonly present prematurely terminated short gagtranscripts, multiply spliced transcripts (msRNAs), andunspliced RNA (usRNA). Levels of usRNA are generallyhigher and therefore better detectable than msRNA. On theother hand, determination of msRNA (e.g., tat/rev; Pasternaket al. 2008) or correctly terminated viral transcripts (usingprimers/probe detecting the polyadenylated tail; Shan et al.2013) better correlates with the ability of the cell to produceinfectious viruses (Bullen et al. 2014; Pasternak et al. 2013).

Based on the PCR detection of tat/rev spliced transcripts, anew quantitative assay allowing single-cell based determina-tion of the inducible viral reservoir called TILDA (Tat/rev-induced limiting dilution assay) was recently presented. The

advantage of this assay is use of only 10 ml of blood, seriallimiting dilutions allowing detection of even single positivecell, and a good correlation with qVOA (Procopio et al. 2015).

A disadvantage of all PCR-based assays is their inability todifferentiate between RNA that might remain retained in thenucleus and for this or other reasons not to be translated into aprotein and RNA giving rise to HIV proteins that could bepresented on the cell surface or constitute new virions (Lassenet al. 2006). Therefore, determination of cell-free RNA(cfRNA) or p24 Ag in culture supernatant better estimatesvirion production. On the other hand, proportion of releasedvirions and detectable cfRNA is much lower in comparisonwith caRNA (1.5 and 7 %, respectively; Cillo et al. 2014).

This problem can be partially solved also by a new assaytermed Prime Flow RNA that combines cell-based detectionof proteins with antibodies and detection of intracellular RNAwith specific probes. It was reported to detect one infected cellin 104–105 cells (comparable numbers are found in peripheralblood of patients on cART) (Romerio and Zapata 2015).

Apparently, highly sensitive assays for determination of HIVproteins in culture supernatant are necessary for a better assess-ment of the efficiency of LRA. One such fully automated assaybased on a Quanterix Simoa technology could detect low levelsof p24 Ag in cell lysates (3 pg/ml; Howell et al. 2015). Anotherapproach might employ TaqMan chemistry-based protein as-says (https://www.thermofisher.com/cz/en/home/life-science/pcr/real-time-pcr/real-time-pcr-applications/real-time-pcr-protein-analysis/protein-expression-taqman-assays.html#workflow).

In summary, assays based on virus outgrowth are very la-borious and generally underestimate the size of the latent poolas their efficiency can be increased by repeated cycles of in-duction of reactivation. On the other hand, technically easierPCR-based detection of HIV-1 prDNA, or caRNA, overesti-mates the size of the latent reservoir, as most of viral DNA ismutated or not transcribed for unknown reasons, while not allRNA transcripts yield functional viral proteins and/or virions.Detection of virion RNA, cfRNA, or viral proteins in culturesupernatant is more accurate but less sensitive due to lowerlevels of these products. Highly sensitive assays for determi-nation of viral proteins are therefore needed.

Mechanisms of establishment and maintenanceof the latent reservoir

There are two types of HIV latency. A pre-integration latencythat occurs after infection of non-permissive cells, which isshort-lived as unintegrated viral DNA is recognized by cyto-plasmic DNA sensors like cGAS or IFI-16 leading to activa-tion of interferon and inflammasome responses (Gao et al.2013; Thompson et al. 2014; Yoh et al. 2015). On the otherhand, the post-integration latency occurring in cells that

Folia Microbiol

Page 7: Current views on HIV-1 latency, persistence, and cure · et al. 2009; Fukazawa et al. 2015; Kim and Siliciano 2016; Van Lint et al. 2013).Additionally,thelatentreservoirisread-ily

become quiescent after the infection is the cause of virus per-sistence. The latent reservoir is established very early duringthe HIV-1 infection and its size can be limited by an earlyintroduction of cART. It consists of both T cells and myeloidcells (Marban et al. 2007; Wires et al. 2012), and it is verystable with half-life determined by qVOA of about 44 months(Crooks et al. 2015; Siliciano et al. 2003).

A key determinant of the future fate of the HIV-1 provirusis the site of its integration. Most commonly, sites of HIV-1insertion are found in intragenic regions of actively tran-scribed genes (Lewinski et al. 2006; Serrao and Engelman2016). Perhaps for this reason, the majority of repressed butinducible proviruses are also located within the introns of theexpressed genes (Lewinski et al. 2005; Shan et al. 2011). Theestablishment of HIV latency is thus regulated independentlyof the control of expression of the target host genes (Mbonyeand Karn 2014).

Insertion of the HIV-1 provirus in the actively transcribedgenes may result in transcriptional interference, contributing tothe regulation of HIV-1 latency. Divergent orientation of thecellular promoter and viral LTR can lead to the lack of recruit-ment of transcription factors, while convergent promoters maylead to a collision of the transcription machinery and prematuretermination of HIV-1 transcription. Parallel orientation of theHIV LTR located downstream of the cellular promoter can leadto the viral promoter occlusion by a read-through transcriptionfrom the cellular gene, displacing key transcription factors onthe HIV LTR (Han et al. 2008; Lenasi et al. 2011). In latentlyinfected cells, a preference for a parallel orientation of the pro-moters was observed, while there was no preference in acutelyor persistently infected cells, suggesting that transcriptional in-terference may be one of the important factors in the establish-ment and maintenance of HIV-1 latency (Shan et al. 2011).

HIV-1 itself does not encode any specific transcription re-pressors, but high levels of HIV-1 transcription activator Tatand its interactions with a cellular cofactor p-TEFb, resolvingpromoter-proximal pausing of RNA polymerase II (RNAP II)are absolutely critical for the provirus expression (Fig. 1;Yamada et al. 2006). In the absence of Tat-pTEFb complex,transcription efficiency decreases drastically and only shortGag transcripts are generated (Kao et al. 1987; Lassen et al.2004; Price 2000). Transcriptional silencing of the provirusesthus results from a series of epigenetic and non-epigeneticchanges occurring at the promoter region and from processesduring the transcription initiation and elongation phases thatdecrease levels of Tat and availability and/or binding of cel-lular factors (Mbonye and Karn 2014).

In the activated CD4+ T cells, which are productively in-fected most often, the intracellular milieu with high levels ofcellular transcription factors, namely transcription initiationfactors NF-κB, nuclear factor of activated T cells (NFAT),and AP-1, drives HIV expression (Mbonye and Karn 2014).However, as the host cell returns to the resting memory

phenotype, cytoplasmic sequestration of these factors causesa significant decrease of transcription initiation at the HIVLTR and allows transcriptional silencing of the provirus, pos-sibly with help of transcriptional inhibitors (Bodor 2006; Heand Margolis 2002; Tyagi and Karn 2007).

The chromatin structure of the proviral 5′ LTR is a criticalparameter in control of HIVexpression. Regardless of the siteof insertion, 5′ HIV LTR is occupied by nucleosomes Nuc-0and Nuc-1 in specific positions at the start site, imposing ablock on RNAP II initiation (Verdin et al. 1993). Several neg-ative DNA-binding factors (e.g., CBF1, YY1, LSF, BRD2,p50 homodimer; He and Margolis 2002; Karn 2013; Tyagiand Karn 2007; Williams et al. 2006) then facilitate recruitingof other repressor complexes and histone- and DNA-modifying enzymes at both core promoter and enhancer re-gions. Histones of the nucleosomes at the 5′ LTR of silentproviruses are deacetylated and trimethylated, which are fea-tures of the repressive heterochromatin. Specifically, atrimethyl mark on histone H3 lysine 27 (H3K27me3), gener-ated through the action of the polycomb repressive complexPRC2, is a mark of the facultative heterochromatin responsi-ble for reversible silencing of various inducible genes andcontributes to viral quiescence, including HIV-1. On the otherhand, H3K9me2/3 is linked with the formation of the consti-tutive heterochromatin during development as well as with theestablishment of HIV-1 latency (Friedman et al. 2011;Maricato et al. 2015; Matsuda et al. 2015). Further, histonemethyltransferases (HMTs) can be found associated with thelatent proviral LTR (du Chene et al. 2007; Friedman et al.2011; Imai et al. 2010; Keedy et al. 2009; Lusic et al. 2013).Namely, a dominant HMT EZH2 constitutes part of the PRC2complex which serves as a binding platform for additionalchromatin-modifying enzymes, histone deacetylases(HDACs), and DNA methyltransferases (DNMTs)(Chenget al. 2011; Friedman et al. 2011; Tae et al. 2011; Vire et al.2006). Methylation of DNA (CpG islands; Bednarik et al.1990) at transcription start site has been suggested to be themost stable modification of the latent provirus LTR. It mightstabilize DNA and prevent provirus reactivation (Blazkovaet al. 2009; Kauder et al. 2009). Lately, relative frequency ofproviruses with a higher LTR DNA methylation was sug-gested to be increased by a prolonged ARV treatment or mul-tiple rounds of reactivation (Trejbalova et al. 2016).

An additional block in HIVexpression may consist in post-transcriptional processes inhibiting HIV-1 protein expression.Namely, both unspliced and spliced HIV-1 transcripts may beretained in nuclei. The export of HIV-1 transcripts is supportedby binding of Rev to Rev response element (RRE) present inpartially spliced and unspliced genomic HIV-1 RNAs and bythe interactions with exportin 1 (Crm-1), a nuclear export fac-tor. Further association with polypyrimidine tract-binding pro-tein (PTB) and related factors seems to affect the export effi-ciency. Thus, unavailability of either factor may promote the

Folia Microbiol

Page 8: Current views on HIV-1 latency, persistence, and cure · et al. 2009; Fukazawa et al. 2015; Kim and Siliciano 2016; Van Lint et al. 2013).Additionally,thelatentreservoirisread-ily

retention of HIV-1 RNAs in the nucleus (Kula et al. 2013;Lassen et al. 2004; Lassen et al. 2006; Zolotukhin et al. 2003).

Further, several cellular miRNAs have been reported tomodulate HIV-1 expression by targeting essential cellular co-factors involved in HIV-1 transcription like PCAF and cyclinT1. The former is targeted bymiR175p andmiR-20awhile thelatter by miR27b and miR198 (Sung and Rice 2009; Tribouletet al. 2007). There are also several cellular miRNAs (e.g.,miR-28, miR-125b, miR-150, miR-223, and miR-382) thatrecognize the 3′-end of HIV-1 mRNAs, repressing their ex-pression in resting CD4+ T cells (Huang et al. 2007). Finally,there are miRNAs derived from viral sequences (vmiRNAs),namely from TAR and Nef (miRTAR5p/3p and miRN367,respectively; Bennasser et al. 2004; Klase et al. 2007;Omoto et al. 2004; Schopman et al. 2012). Both cellular andviral miRNAs may cause HIV-1 RNA degradation or ineffi-cient expression of HIV-1 proteins.

Mechanisms of reactivation

The reactivation of the functional, inducible latent provirusdepends on the chromatin status and availability of the cellulartranscription factors (Fig. 1). Upon appropriate stimulationand nuclear translocation, NF-κB and NFAT can bind toHIV LTR if the chromatin landscape of the promoter regionis favorable (Bhatt and Ghosh 2014; Lusic et al. 2013; Ott andVerdin 2013). The accessibility of the HIV LTR can be affect-ed by methylation of CpG islands (Bednarik et al. 1990) orbinding of other transcription factors like Sp1 that are able topromote the chromatin configuration favorable for binding ofthe main transcription factors. In fact, Sp1 is required for theformation of the pre-initiation complex and interacts withNF-κB (Perkins et al. 1993). NF-κB and NFAT probably bindin a mutually exclusive, possibly sequential way, as their HIV-1 LTR-binding sites overlap (Giffin et al. 2003; Mbonye andKarn 2014). NF-κB is found in cell lines and primary naïve Tcells, while NFAT is typically present in memory T cells(Dienz et al. 2007). In primary memory T cells, NFAT andLck are required for optimal latent virus reactivation andHIV-1 can be activated in an NF-kB-independent way bytranscription factor DVII-Ets-1, without causing significantT cell activation (Bosque et al. 2011; Bosque and Planelles2009; Yang et al. 2009a). On the other hand, NFAT is dispens-able in Jurkat cell models. NF-κB is commonly activated byPMA or TNF-α, while NFAT is activated by calcium/calcineurin signaling (Bosque and Planelles 2009; Chanet al. 2013; Kim et al. 2011).

Both NF-κB and NFAT recruit CBP/p300 and other histoneacetyltransferases (HATs) (Garcia-Rodriguez and Rao 1998) tofurther acetylate Nuc-1 and to attract SWI/SNF chromatin re-modeling complex. After the minimal initiation andtranscription through the TAR element, RNAP II pauses. If

Tat and p-TEFb, composed of CDK9 and cyclin T1(Herrmann and Rice 1995; Wei et al. 1998) are available tobind and form a complex with TAR RNA hairpin and the tran-scription machinery, kinase activity of p-TEFb mediates phos-phorylation of negative elongation factors (DSIF and NELF)and of RNAP II and allows formation of a superelongationcomplex and continuation of transcription further into the elon-gation phase (Fig. 1). Thus, Tat transactivation and its interac-tion with p-TEFb is absolutely necessary for the efficienttranscription elongation of the HIV provirus. When notcomplexed with Tat, pTEFb is sequestered and held inactivein the transcriptionally inactive 7SK RNP complex containing7SK small nuclear RNA (7SK snRNA), inhibitory factorHEXIM1 and RNA binding proteins LARP7 and MePCE.Yet, Tat may be out-competed by BRD4 in binding top-TEFb, leading to p-TEFb targeting to transcription of cellulargenes (Mbonye and Karn 2014).

Besides this conventional view of events underlying theHIV-1 reactivation, it has been suggested that the decisionbetween HIV-1 replication and latency can be determined bya stochastic noise in gene expression similarly as in other cell-fate decisions. In this view, the probability of HIV-1 reactiva-tion is increased by a higher rate and variation of fluctuationsin HIV-1 gene expression and further stimulated by Tat-mediated positive feedback mechanism (Dar et al. 2014; Hoet al. 2013; Weinberger 2015; Weinberger et al. 2005;Weinberger et al. 2008).

Latency reversal and purging HIV-1

Latently infected cells represent the major barrier to HIV-1cure (Donahue and Wainberg 2013). The major reservoir isconsidered to reside in resting memory T cells, and therefore,this population is in the focus of most efforts to decrease thesize of the latent reservoir (Bruner et al. 2015).

The initial attempts to eradicate HIV-1 latently infectedcells were first described by Fauci et al. (Chun et al. 1998;Chun and Fauci 1999). Later, the term shock and kill strategywas introduced (Archin et al. 2012; Deeks 2012). In short, itconsists in the attempts to reactivate a dormant provirus silent-ly present in latently infected cells, namely long-livedmemoryCD4 Tcells, that would lead to death of the cells harboring thelatent provirus and decrease the size of the latent pool in thepresence of cART. Originally, it was assumed that virus reac-tivation could be achieved with a single agent, namely HDACinhibitors (HDACi) or PKC inducers, and that the replicationof the reactivated virus and the virus-induced cytopathic ef-fects would be sufficient to kill the host cells and thus decreasethe size of the latent reservoir. Today, it is largely accepted thatcombinations of two or more agents with different mechanismof action together with an additional stimulation of anti-HIVimmune responses would be necessary. Namely, improvement

Folia Microbiol

Page 9: Current views on HIV-1 latency, persistence, and cure · et al. 2009; Fukazawa et al. 2015; Kim and Siliciano 2016; Van Lint et al. 2013).Additionally,thelatentreservoirisread-ily

of cytotoxic responses using engineered dual-affinity (DART)and broadly specific antibodies (bNAbs) toward HIV-1 areintensively investigated (Caskey et al. 2015; Sloan et al.2015). Still, there are doubts if the HIV cure can be everachieved because of unknown fraction of the cells remain-ing refractory to the reactivation strategies. Further, it hasbeen suggested that a prolonged antiretroviral treatmenttogether with random or intentional cycles of reactivationmight lead to an increased methylation of HIV-LTR andthus stabilization of the latent provirus (Trejbalova et al.2016). Therefore, another logical approach would be toinhibit the spontaneous reactivation of latent HIV-1(Mousseau et al. 2015).

Activity of LRA is usually identified in vitro in tissue culturescreens involving various cell lines and primary cells. The effi-ciency of the selected compounds must then be verified in vivo.Most intensively investigated LRA, namely those in clinicaltrials, involve compounds in use or under development fortreatment of various cancers and other diseases. These are es-pecially HDACi and PKC agonists (NF-κB inducers). Thereare 11 HDACs subdivided in four classes (Mottamal et al.2015; Wang et al. 2009). Particularly, HDACs 1–3 (class I)seem to be important in maintaining HIV latency (Keedyet al. 2009). HDACi non-specifically activate transcription ofmany genes by increasing acetylation of the promoter regions,including HIV LTR. Increased acetylation should modify chro-matin status and allow for binding of transcription factors,namely NF-κB and NFAT. Among these, vorinostat (SAHA)has been the most extensively explored, but newer compoundswith higher potency like givinostat, panobinostat, orromidepsin seem more promising (Banga et al. 2016). Theability of individual HDACi alone to induce HIV-1 reactivationex vivo, in cells isolated fromHIV-1-infected patients on cART,apparently depends on specific experimental conditions as wellas on patients’ history, as not all attempts ex vivo were success-ful (Blazkova et al. 2012; Bullen et al. 2014; Rasmussen et al.2013). However, lately, vorinostat, panobinostat, romidepsin,and namely givinostat were found effective in a modifiedVOA employing a prolonged or repeated treatment with indi-vidual HDACi (Banga et al. 2016), repeating the results ofclinical studies, in which vorinostat and some other HDACiwere able to increase levels of caRNA (Archin et al. 2014a;Rasmussen et al. 2014; Sogaard et al. 2015). Nevertheless inthese studies, the size of the latent reservoir was not founddecreased in vivo, suggesting that combination with otheragents or strategies increasing immunological killing of theinfected cells would be necessary (Rasmussen et al. 2013).Other classes of chromatin-modifying enzyme inhibitors in-clude inhibitors of histone methyltransferases (HMTi) likechaetocin, BIX-01294 or GSK343, or inhibitors of DNAmeth-yltransferases (DNMTi) like 5-aza-2′deoxycytidine. However,they are more likely to be effective in combination with otherLRA (Kumar et al. 2015).

The other important classes of LRA represent PKC ago-nists that induce activation and nuclear translocation NF-κBand p-TEFb. They can also trigger activation of MAPK andnuclear translocation of AP1. The natural PKC inducer effec-tive in HIV-1 reactivation is TNF-α, a cytokine found in-creased in untreated HIV-1 infection. However, due to itspro-inflammatory pleiotropic effects, its use as a LRAin vivo cannot be considered. Similarly, the other very effec-tive agent of this class, phorbol myristate acetate (PMA), can-not be considered for therapy as it reveals a strong tumorigenicpotential (Kumar et al. 2015). However, newer PKC inducersinclude non-tumorigenic phorbol ester prostratin (Biancottoet al. 2004; Kulkosky et al. 2001), macrolide lactonebryostatin-1 (Mehla et al. 2010), or diterpenoids ingenol Band ingenol-3-angelate (Jiang et al. 2014; Jiang et al. 2015).PKC inducers downregulate expression of cell surface recep-tors CD4, CXCR4, or CCR5 in uninfected cells, thus limitingthe spread of the newly released virus (Hezareh et al. 2004;Jiang et al. 2014; Mehla et al. 2010). Further, PKC was report-ed to phosphorylate HEXIM1, suggesting that PKC inducersmight affect also this inhibitory protein (Fujinaga et al. 2012).Hexim phosphorylation is commonly mediated by AKT ki-nase that can be stimulated by HMBA (Contreras et al. 2007),leading to the release of p-TEFb from the inhibitory complex7SK RNP. Yet another compound affecting availability of p-TEFb is JQ1, bromodomain inhibitor affecting factors BRD2and BRD4 (Boehm et al. 2013).

Disulfiram, the inhibitor of acetaldehyde dehydrogenaseused in therapy of alcohol abuse, induces degradation ofPTEN, again allowing AKT-mediated phosphorylation ofHEXIM1 (Doyon et al. 2013; Xing et al. 2011). This compoundhas been described as an effective LRA in vitro in a relativelyartificial model of primary CD4+ T cells immortalized with theBcl-2 protooncogene. However, the expectations of its potencywere not fulfilled as there was no significant effect on the size ofthe latent reservoir found in vivo (Spivak et al. 2014).

As mentioned above, combinations of several LRA arelikely to act more efficiently and cause fewer negative sideeffects. The examples of such combinations are chromatin-remodeling compounds, namely HDACi, together with differ-ent inducers of transcription. Chromatin remodeling com-pounds can be apparently considered as noise enhancers thatare relatively ineffective in HIV-1 reactivation alone, whilethey reveal a synergismwith real transcriptional activators thatare able to increase HIV-1 expression alone (e.g., PKC in-ducers) (Dar et al. 2014; Kumar et al. 2015).

Apart from the mainstream studies, there are many otherapproaches toward latency reversal and HIV cure. Of these,use of a pro-oxidant gold-based drug Auranofin is of interestas it was shown to induce a partially selective killing of TCMand TTM CD4+ T cells, the main reservoir cells containingthe latent HIV-1 (Lewis et al. 2011). Consequently, it wasdemonstrated that TCM and TTM CD4+ T lymphocytes are

Folia Microbiol

Page 10: Current views on HIV-1 latency, persistence, and cure · et al. 2009; Fukazawa et al. 2015; Kim and Siliciano 2016; Van Lint et al. 2013).Additionally,thelatentreservoirisread-ily

more susceptible to the redox stress and apoptosis (Chirulloet al. 2013).

Our laboratory has demonstrated independently thatNormosang, a hemin containing compound used to treat acutehepatic porphyria, could strongly potentiate reactivation of thelatent provirus induced by PKC inducers like PMA, TNF-α,prostratin or bryostatin-1, while it inhibits HIV-1 replicationduring the acute infection through its effect on reversetranscription. The stimulatory effects of Normosang are medi-ated by a heme/iron-mediated Fenton reaction resulting in theincreased redox stress, while there was no effect on the activa-tion status of the cells (Melkova et al. 2015a; Melkova et al.2015b; Shankaran et al. 2011). Based on our results, wepropose a model in which redox-modulating agents inducechromatin remodeling, affect binding of specific transcriptionfactors to HIV-LTR, and potentiate HIV-1 expression inducedby PKC or other inducers (Melkova et al. 2015a; Melkova et al.2015b).

Historically, there is a case of one HIV+ patient that wasadministered Normosang and consequently remained p24-negative for several months (Pavel Martasek, FacultyGeneral Hospital Prague, personal communication). We sug-gest that the outcome in this particular patient resulted fromthe inhibition of reverse transcription by heme (Levere et al.1991) together with a short-term reactivation and death of theinfected cells due to heme/iron-mediated redox stress.Consequently, a stable heme degradation product, antioxidantbilirubin, might havemediated prolonged inhibitory effects onHIV-1 reactivation (Melkova et al. 2015a; Melkova et al.2015b). This scenario would be compatible with the hypoth-esis of Chirullo et al. (2013) that auranofin decreases numbersof latently infected TCM and TTM CD4+ T cells by its pro-oxidant effects and thus reduces size of the HIV latent pool.

The idea of possible involvement and use of redox stress topurge the latent reservoir is further supported by work of(Iordanskiy et al. 2015) showing that ionizing radiation alonewas sufficient to activate the HIV-1 LTR and to effectively killthe infected T cells. Consequently, this group proposed that alow-dose ionizing radiation could be used therapeutically toreactivate and kill latently infected reservoir cells (Iordanskiyand Kashanchi 2016). Additionally, in the Berlin patient, theonly known case of HIV-1 cure, high-dose irradiationwith bonemarrow transplantation from a donor harboring CCR5Δ32mu-tation with missing CCR5 expression was used, while irradia-tion was omitted in Boston patients in which virus reboundoccurred (Henrich et al. 2013; Hutter et al. 2015).

Finally, any common infection involves increased genera-tion of reactive oxygen and nitrogen species. Co-infections arewell-known to increase HIV-1 replication and promote itsspread (Modjarrad and Vermund 2010), while decreased levelsof GSH, indicator of an increased redox stress, have beendescribed early in HIV infection (Pace and Leaf 1995).Reactive oxygen and nitrogen species were shown to stimulate

activation of the redox-sensitive transcription factor NF-κBand LTR-driven expression of reporter genes (Jimenez et al.2001; Melkova et al. 2000; Pyo et al. 2008). Thus, increasedgeneration of free radicals is likely to be helpful in the attemptsto eliminate the latent HIV-1 in the presence of cART.

A major concern when considering testing of LRA in vivois the induction of immune hyperstimulation upon reactivationof the latent HIV-1 and development of a harmful cytokinestorm. On the other hand, it is not clear if any significantlatency reversal is achievable without increased cytokinelevels (Marsden et al. 2015). However, specific anti-HIV im-mune responses are generally hampered or dysfunctional.Further, it was shown that both HDACi (romidepsin,panobinostat) and PKC agonists (prostratin and bryostatin-1,but not ingenol B) inhibited HIV-specific T cell proliferation(Clutton et al. 2015).

Based on clinical studies, it has been widely accepted thatstimulation or improved function of immune responses in ad-dition to LRAwould be necessary in order to achieve death ofreactivated cells and thus decrease the size of the latent reser-voir. Synergistic effects of different agents should also de-crease the doses and/or negative side effects of individualcompounds.

Summary/conclusions

In conclusion, in view of the currently described sources of theremaining low-level HIV-1 replication in the presence of sup-pressive cART, additional mechanisms and processes promot-ing HIV-1 persistence remain to be identified. Any attempt toachieve HIV-1 cure by latency reversal should involve com-bination of several LRA with different mechanism of actiontogether with stimulation or improvement of immune re-sponses toward infected cells. To validate the efficiency ofindividual approaches, development of sufficiently sensitiveand specific methods allowing accurate determination of thesize of the latent reservoir and its changes is necessary.

Acknowledgments We are grateful to Dr. Ivan Hirsch for helpful dis-cussions and comments. The work of P.S. and M.M. was performed inpartial fulfillment of the requirements for Ph.D. degree at the FirstMedical Faculty, Charles University. The work was supported by theGrant Agency of the Ministry of Health of the Czech Republic – projectNo. NT/14135-3.

References

Archin NM et al (2012) Administration of vorinostat disrupts HIV-1latency in patients on antiretroviral therapy. Nature 487:482–485.doi:10.1038/nature11286

Archin NM et al (2014a) HIV-1 expression within resting CD4+ T cellsafter multiple doses of vorinostat. The Journal of infectious diseases210:728–735. doi:10.1093/infdis/jiu155

Folia Microbiol

Page 11: Current views on HIV-1 latency, persistence, and cure · et al. 2009; Fukazawa et al. 2015; Kim and Siliciano 2016; Van Lint et al. 2013).Additionally,thelatentreservoirisread-ily

Archin NM, Sung JM, Garrido C, Soriano-Sarabia N, Margolis DM(2014b) Eradicating HIV-1 infection: seeking to clear a persistentpathogen nature reviews. Microbiology 12:750–764. doi:10.1038/nrmicro3352

Avalos CR et al (2016) Quantitation of productively infected monocytesand macrophages of SIV-infected macaques. J Virol. doi:10.1128/JVI.00290-16

Banga R, Procopio FA, CavassiniM, PerreauM (2016) In vitro reactivationof replication-competent and infectious HIV-1 by histone deacetylaseinhibitors. J Virol 90:1858–1871. doi:10.1128/JVI.02359-15

Bednarik DP, Cook JA, Pitha PM (1990) Inactivation of the HIV LTR byDNA CpG methylation: evidence for a role in latency. EMBO J 9:1157–1164

Bennasser Y, Le SY, Yeung ML, Jeang KT (2004) HIV-1 encoded can-didate micro-RNAs and their cellular targets. Retrovirology 1:43.doi:10.1186/1742-4690-1-43

Bhatt D, Ghosh S (2014) Regulation of the NF-kappaB-mediated tran-scription of inflammatory genes. Front Immunol 5:71. doi:10.3389/fimmu.2014.00071

Biancotto A et al (2004) Dual role of prostratin in inhibition of infectionand reactivation of human immunodeficiency virus from latency inprimary blood lymphocytes and lymphoid tissue. J Virol 78:10507–10515. doi:10.1128/JVI.78.19.10507-10515.2004

Blazkova J et al (2009) CpG methylation controls reactivation of HIVfrom latency. PLoS Pathog 5:e1000554. doi:10.1371/journal.ppat.1000554

Blazkova J et al (2012) Paucity of HIV DNA methylation in latentlyinfected, resting CD4+ T cells from infected individuals receivingantiretroviral therapy. J Virol 86:5390–5392. doi:10.1128/JVI.00040-12

BlumKS, Pabst R (2007) Lymphocyte numbers and subsets in the humanblood. Do they mirror the situation in all organs? Immunol Lett 108:45–51. doi:10.1016/j.imlet.2006.10.009

Bodor J (2006) Regulation of HIV-1 and IL-2 transcription by inducibleCAMP early repressor (ICER. Retrovirology 3(Suppl 1):S81

Boehm D et al (2013) BET bromodomain-targeting compounds reacti-vate HIV from latency via a Tat-independent mechanism. Cell Cycle12:452–462. doi:10.4161/cc.23309

Bosque A, Planelles V (2009) Induction of HIV-1 latency and reactiva-tion in primary memory CD4+ T cells. Blood 113:58–65.doi:10.1182/blood-2008-07-168393

Bosque A, Famiglietti M, Weyrich AS, Goulston C, Planelles V (2011)Homeostatic proliferation fails to efficiently reactivate HIV-1 latent-ly infected central memory CD4+ Tcells. PLoS Pathog 7:e1002288.doi:10.1371/journal.ppat.1002288

Brooks DG, Kitchen SG, Kitchen CM, Scripture-Adams DD, Zack JA(2001) Generation of HIV latency during thymopoiesis. Nat Med 7:459–464. doi:10.1038/86531

Bruner KM, Hosmane NN, Siliciano RF (2015) Towards an HIV-1 cure:measuring the latent reservoir. Trends Microbiol 23:192–203.doi:10.1016/j.tim.2015.01.013

Bullen CK, Laird GM, Durand CM, Siliciano JD, Siliciano RF (2014)New ex vivo approaches distinguish effective and ineffective singleagents for reversing HIV-1 latency in vivo. Nat Med 20:425–429.doi:10.1038/nm.3489

Buzon MJ et al (2014) HIV-1 persistence in CD4+ T cells with stem cell-like properties. Nat Med 20:139–142. doi:10.1038/nm.3445

Carter CC, Onafuwa-Nuga A, McNamara LA, Riddell J, Bixby D,Savona MR, Collins KL (2010) HIV-1 infects multipotent progeni-tor cells causing cell death and establishing latent cellular reservoirs.Nat Med 16:446–451. doi:10.1038/nm.2109

CaskeyM et al (2015) Viraemia suppressed in HIV-1-infected humans bybroadly neutralizing antibody 3BNC117. Nature 522:487–491.doi:10.1038/nature14411

Chan JK, Bhattacharyya D, Lassen KG, Ruelas D, Greene WC (2013)Calcium/calcineurin synergizes with prostratin to promote NF-

kappaB dependent activation of latent HIV. PLoS One 8:e77749.doi:10.1371/journal.pone.0077749

Chege D et al (2011) Sigmoid Th17 populations, the HIV latent reservoir,and microbial translocation in men on long-term antiretroviral ther-apy. AIDS 25:741–749. doi:10.1097/QAD.0b013e328344cefb

Cheng AS et al (2011) EZH2-mediated concordant repression of Wntantagonists promotes beta-catenin-dependent hepatocarcinogenesis.Cancer Res 71:4028–4039. doi:10.1158/0008-5472.CAN-10-3342

Chirullo B et al (2013) A candidate anti-HIV reservoir compound,auranofin, exerts a selective ‘anti-memory’ effect by exploiting thebaseline oxidative status of lymphocytes. Cell Death Dis 4:e944.doi:10.1038/cddis.2013.473

Chomont N et al (2009) HIV reservoir size and persistence are driven byT cell survival and homeostatic proliferation. Nat Med 15:893–900.doi:10.1038/nm.1972

Choudhary SK, Archin NM, Cheema M, Dahl NP, Garcia JV, MargolisDM (2012) Latent HIV-1 infection of resting CD4(+) T cells in thehumanized Rag2(−)/(−) gamma c(−)/(−) mouse. J Virol 86:114–120. doi:10.1128/JVI.05590-11

Chun TW, Fauci AS (1999) Latent reservoirs of HIV: obstacles tothe eradication of virus. Proc Natl Acad Sci U S A 96:10958–10961

Chun TW, Engel D, Mizell SB, Ehler LA, Fauci AS (1998) Induction ofHIV-1 replication in latently infected CD4+ T cells using a combi-nation of cytokines. J Exp Med 188:83–91

Cillo AR, Sobolewski MD, Bosch RJ, Fyne E, Piatak M Jr, Coffin JM,Mellors JW (2014) Quantification of HIV-1 latency reversal in restingCD4+Tcells from patients on suppressive antiretroviral therapy. ProcNatl Acad Sci U S A 111:7078–7083. doi:10.1073/pnas.1402873111

Clouse KA et al (1989) Monokine regulation of human immunodeficien-cy virus-1 expression in a chronically infected human T cell clone. JImmunol 142:431–438

Clutton G, Archin N, Xu Y, Margolis DM, Goonetilleke N (2015)Differential effects of HIV latency-reversing agents on T cell phe-notype and function: implications for HIV cure. Journal of VirusEradication 1:8

Contreras X, Barboric M, Lenasi T, Peterlin BM (2007) HMBA releasesP-TEFb fromHEXIM1 and 7SK snRNAvia PI3K/Akt and activatesHIV transcription. PLoS Pathog 3:1459–1469. doi:10.1371/journal.ppat.0030146

Crooks AM et al (2015) Precise quantitation of the latent HIV-1 reservoir:implications for eradication strategies. The Journal of InfectiousDiseases 212:1361–1365. doi:10.1093/infdis/jiv218

Dar RD, Hosmane NN, Arkin MR, Siliciano RF, Weinberger LS (2014)Screening for noise in gene expression identifies drug synergies.Science 344:1392–1396. doi:10.1126/science.1250220

Deeks SG (2012) HIV: shock and kill. Nature 487:439–440. doi:10.1038/487439a

Denton PW et al (2012) Generation of HIV latency in humanized BLTmice. J Virol 86:630–634. doi:10.1128/JVI.06120-11

Dienz O et al (2007) Accumulation of NFAT mediates IL-2 expression inmemory, but not naive, CD4+ T cells. Proc Natl Acad Sci U S A104:7175–7180. doi:10.1073/pnas.0610442104

DoitshG et al (2014) Cell death by pyroptosis drives CD4 T-cell depletionin HIV-1 infection. Nature 505:509–514. doi:10.1038/nature12940

Donahue DA, Wainberg MA (2013) Cellular and molecular mechanismsinvolved in the establishment of HIV-1 latency. Retrovirology 10:11. doi:10.1186/1742-4690-10-11

Doyon G, Zerbato J, Mellors JW, Sluis-Cremer N (2013) Disulfiramreactivates latent HIV-1 expression through depletion of the phos-phatase and tensin homolog. AIDS 27:F7–F11. doi:10.1097/QAD.0b013e3283570620

du Chene I et al (2007) Suv39H1 and HP1gamma are responsible forchromatin-mediated HIV-1 transcriptional silencing and post-integration latency. EMBO J 26:424–435. doi:10.1038/sj.emboj.7601517

Folia Microbiol

Page 12: Current views on HIV-1 latency, persistence, and cure · et al. 2009; Fukazawa et al. 2015; Kim and Siliciano 2016; Van Lint et al. 2013).Additionally,thelatentreservoirisread-ily

Farber D (2015) Tissue localization of human Tcell responses. Journal ofVirus Eradication 1:6

Farber DL, Yudanin NA, Restifo NP (2014) Human memory T cells:generation, compartmentalization and homeostasis nature reviews.Immunology 14:24–35. doi:10.1038/nri3567

Finzi D et al (1997) Identification of a reservoir for HIV-1 in patients onhighly active antiretroviral therapy. Science 278:1295–1300

Finzi D et al (1999) Latent infection of CD4+ T cells provides a mecha-nism for lifelong persistence of HIV-1, even in patients on effectivecombination therapy. Nat Med 5:512–517. doi:10.1038/8394

Fletcher CV et al (2014) Persistent HIV-1 replication is associated withlower antiretroviral drug concentrations in lymphatic tissues. ProcNatl Acad Sci U S A 111:2307–2312. doi:10.1073/pnas.1318249111

Folks TM, Justement J, Kinter A, Dinarello CA, Fauci AS (1987)Cytokine-induced expression of HIV-1 in a chronically infectedpromonocyte cell line. Science 238:800–802

Folks TM, Clouse KA, Justement J, Rabson A, Duh E, Kehrl JH, FauciAS (1989) Tumor necrosis factor alpha induces expression of hu-man immunodeficiency virus in a chronically infected T-cell clone.Proc Natl Acad Sci U S A 86:2365–2368

Friedman J, Cho WK, Chu CK, Keedy KS, Archin NM, Margolis DM,Karn J (2011) Epigenetic silencing of HIV-1 by the histone H3lysine 27 methyltransferase enhancer of Zeste 2. J Virol 85:9078–9089. doi:10.1128/JVI.00836-11

Fujinaga K, BarboricM, Li Q, Luo Z, Price DH, Peterlin BM (2012) PKCphosphorylates HEXIM1 and regulates P-TEFb activity. NucleicAcids Res 40:9160–9170. doi:10.1093/nar/gks682

Fukazawa Y et al (2015) B cell follicle sanctuary permits persistent pro-ductive simian immunodeficiency virus infection in elite controllers.Nat Med 21:132–139. doi:10.1038/nm.3781

Gao D et al (2013) Cyclic GMP-AMP synthase is an innate immunesensor of HIV and other retroviruses. Science 341:903–906.doi:10.1126/science.1240933

Garcia-Rodriguez C, Rao A (1998) Nuclear factor of activated T cells(NFAT)-dependent transactivation regulated by the coactivatorsp300/CREB-binding protein (CBP). J Exp Med 187:2031–2036

Gardner MB, Luciw PA (2008) Macaque models of human infectiousdisease. ILAR Journal/National Research Council, Institute ofLaboratory Animal Resources 49:220–255

Giffin MJ, Stroud JC, Bates DL, von Koenig KD, Hardin J, Chen L(2003) Structure of NFAT1 bound as a dimer to the HIV-1 LTRkappa B element. Nat Struct Biol 10:800–806. doi:10.1038/nsb981

Gludish DW,MwandumbaHC, Jambo KC, Amie SM, Russel DG (2015)The human lung is a site of productive HIV infection during long-termART: novel tools to study ART-durable HIV reservoirs. Journalof Virus Eradication 1:7

Gondois-Rey F et al (2006) R5 variants of human immunodeficiencyvirus type 1 preferentially infect CD62L- CD4+ T cells and arepotentially resistant to nucleoside reverse transcriptase inhibitors. JVirol 80:854–865. doi:10.1128/JVI.80.2.854-865.2006

Han Yet al (2008) Orientation-dependent regulation of integrated HIV-1expression by host gene transcriptional readthrough. Cell HostMicrobe 4:134–146. doi:10.1016/j.chom.2008.06.008

HeG,Margolis DM (2002) Counterregulation of chromatin deacetylationand histone deacetylase occupancy at the integrated promoter ofhuman immunodeficiency virus type 1 (HIV-1) by the HIV-1 repres-sor YY1 and HIV-1 activator Tat. Mol Cell Biol 22:2965–2973

Henrich TJ et al (2013) Long-term reduction in peripheral blood HIV type1 reservoirs following reduced-intensity conditioning allogeneicstem cell transplantation. The Journal of Infectious Diseases 207:1694–1702. doi:10.1093/infdis/jit086

Herrmann CH, Rice AP (1995) Lentivirus Tat proteins specifically asso-ciate with a cellular protein kinase, TAK, that hyperphosphorylatesthe carboxyl-terminal domain of the large subunit of RNA polymer-ase II: candidate for a Tat cofactor. J Virol 69:1612–1620

Hezareh M et al (2004) Mechanisms of HIV receptor and co-receptordown-regulation by prostratin: role of conventional and novelPKC isoforms. Antiviral Chemistry & Chemotherapy 15:207–222

Hilldorfer BB, Cillo AR, Besson GJ, Bedison MA, Mellors JW (2012)New tools for quantifying HIV-1 reservoirs: plasma RNA singlecopy assays and beyond. Current HIV/AIDS Reports 9:91–100.doi:10.1007/s11904-011-0104-6

Hirsch VM, Olmsted RA, Murphey-Corb M, Purcell RH, Johnson PR(1989) An African primate lentivirus (SIVsm) closely related toHIV-2. Nature 339:389–392. doi:10.1038/339389a0

Ho DD, Neumann AU, Perelson AS, Chen W, Leonard JM, MarkowitzM (1995) Rapid turnover of plasma virions and CD4 lymphocytes inHIV-1 infection. Nature 373:123–126. doi:10.1038/373123a0

Ho YC et al (2013) Replication-competent noninduced proviruses in thelatent reservoir increase barrier to HIV-1 cure. Cell 155:540–551.doi:10.1016/j.cell.2013.09.020

Hofmann H et al (2012) The Vpx lentiviral accessory protein targetsSAMHD1 for degradation in the nucleus. J Virol 86:12552–12560. doi:10.1128/JVI.01657-12

Honeycutt JB, Wahl A, Archin N, Choudhary S, Margolis D, Garcia JV(2013) HIV-1 infection, response to treatment and establishment ofviral latency in a novel humanized Tcell-only mouse (TOM) model.Retrovirology 10:121. doi:10.1186/1742-4690-10-121

Honeycutt JB et al (2016) Macrophages sustain HIV replication in vivoindependently of Tcells. J Clin Invest 126:1353–1366. doi:10.1172/JCI84456

Howell BJ et al (2015) Developing and applying ultrasensitive p24 pro-tein immunoassay for HIV latency. Journal of Virus Eradication 1:5

Huang J et al (2007) Cellular microRNAs contribute to HIV-1 latency inresting primary CD4+ T lymphocytes. Nat Med 13:1241–1247.doi:10.1038/nm1639

Huang Y et al (2016) Antiretroviral drug transporters and metabolic en-zymes in human testicular tissue: potential contribution to HIV-1sanctuary site. J Antimicrob Chemother. doi:10.1093/jac/dkw046

Huet T, Cheynier R, Meyerhans A, Roelants G, Wain-Hobson S (1990)Genetic organization of a chimpanzee lentivirus related to HIV-1.Nature 345:356–359. doi:10.1038/345356a0

Hutter G, Bodor J, Ledger S, BoydM,MillingtonM, TsieM, Symonds G(2015) CCR5 targeted cell therapy for HIV and prevention of viralescape. Viruses 7:4186–4203. doi:10.3390/v7082816

Imai K, Togami H,Okamoto T (2010) Involvement of histone H3 lysine 9(H3K9) methyltransferase G9a in the maintenance of HIV-1 latencyand its reactivation by BIX01294. J Biol Chem 285:16538–16545.doi:10.1074/jbc.M110.103531

Iordanskiy S, Kashanchi F (2016) Potential of radiation-induced cellularstress for reactivation of latent HIV-1 and killing of infected cells.AIDS Res Hum Retrovir 32:120–124. doi:10.1089/AID.2016.0006

Iordanskiy S et al (2015) Therapeutic doses of irradiation activate viraltranscription and induce apoptosis in HIV-1 infected cells. Virology485:1–15. doi:10.1016/j.virol.2015.06.021

Jiang Y, Tian B, Saifuddin M, Agy MB, Emau P, Cairns JS, Tsai CC(2009) RT-SHIV, an infectious CCR5-tropic chimeric virus suitablefor evaluating HIV reverse transcriptase inhibitors in macaquemodels. AIDS Res Ther 6:23. doi:10.1186/1742-6405-6-23

Jiang G et al (2014) Reactivation of HIV latency by a newly modifiedIngenol derivative via protein kinase Cdelta-NF-kappaB signaling.AIDS 28:1555–1566. doi:10.1097/QAD.0000000000000289

Jiang G et al (2015) Synergistic reactivation of latent HIV expression byIngenol-3-Angelate, PEP005, targeted NF-kB signaling in combina-tion with JQ1 induced p-TEFb activation. PLoS Pathog 11:e1005066. doi:10.1371/journal.ppat.1005066

Jimenez JL, Gonzalez-Nicolas J, Alvarez S, FresnoM,Munoz-FernandezMA (2001) Regulation of human immunodeficiency virus type 1replication in human T lymphocytes by nitric oxide. J Virol 75:4655–4663. doi:10.1128/JVI.75.10.4655-4663.2001

Folia Microbiol

Page 13: Current views on HIV-1 latency, persistence, and cure · et al. 2009; Fukazawa et al. 2015; Kim and Siliciano 2016; Van Lint et al. 2013).Additionally,thelatentreservoirisread-ily

Jordan A, Bisgrove D, Verdin E (2003) HIV reproducibly establishes alatent infection after acute infection of T cells in vitro. EMBO J 22:1868–1877. doi:10.1093/emboj/cdg188

Kao SY, Calman AF, Luciw PA, Peterlin BM (1987) Anti-termination oftranscription within the long terminal repeat of HIV-1 by tat geneproduct. Nature 330:489–493. doi:10.1038/330489a0

Karn J (2013) A new BET on the control of HIV latency. Cell Cycle 12:545–546. doi:10.4161/cc.23679

Karpel ME, Boutwell CL, Allen TM (2015) BLT humanized mice as asmall animal model of HIV infection. Current Opinion in Virology13:75–80. doi:10.1016/j.coviro.2015.05.002

Kauder SE, Bosque A, Lindqvist A, Planelles V, Verdin E (2009)Epigenetic regulation of HIV-1 latency by cytosine methylation.PLoS Pathog 5:e1000495. doi:10.1371/journal.ppat.1000495

Keedy KS, Archin NM, Gates AT, Espeseth A, Hazuda DJ,Margolis DM(2009) A limited group of class I histone deacetylases acts to represshuman immunodeficiency virus type 1 expression. J Virol 83:4749–4756. doi:10.1128/JVI.02585-08

Kim M, Siliciano RF (2016) Reservoir expansion by T-cell proliferationmay be another barrier to curing HIV infection. Proc Natl Acad SciU S A 113:1692–1694. doi:10.1073/pnas.1600097113

Kim YK, Mbonye U, Hokello J, Karn J (2011) T-cell receptor signalingenhances transcriptional elongation from latent HIV proviruses byactivating P-TEFb through an ERK-dependent pathway. J Mol Biol410:896–916. doi:10.1016/j.jmb.2011.03.054

Kiselinova M, Pasternak AO, De Spiegelaere W, Vogelaers D, BerkhoutB, Vandekerckhove L (2014) Comparison of droplet digital PCRand seminested real-time PCR for quantification of cell-associatedHIV-1 RNA. PLoS One 9:e85999. doi:10.1371/journal.pone.0085999

Klase Z et al (2007) HIV-1 TAR element is processed by dicer to yield aviral micro-RNA involved in chromatin remodeling of the viralLTR. BMC Mol Biol 8:63. doi:10.1186/1471-2199-8-63

Kula A, Gharu L, Marcello A (2013) HIV-1 pre-mRNA commitment torev mediated export through PSF and Matrin 3. Virology 435:329–340. doi:10.1016/j.virol.2012.10.032

Kulkosky J, Culnan DM, Roman J, Dornadula G, Schnell M, Boyd MR,Pomerantz RJ (2001) Prostratin: activation of latent HIV-1 expres-sion suggests a potential inductive adjuvant therapy for HAART.Blood 98:3006–3015

Kumar A, Darcis G, Van Lint C, Herbein G (2015) Epigenetic control ofHIV-1 post integration latency: implications for therapy. ClinicalRpigenetics 7:103. doi:10.1186/s13148-015-0137-6

Lassen KG, Bailey JR, Siliciano RF (2004) Analysis of human immuno-deficiency virus type 1 transcriptional elongation in resting CD4+ Tcells in vivo. J Virol 78:9105–9114. doi:10.1128/JVI.78.17.9105-9114.2004

Lassen KG, Ramyar KX, Bailey JR, ZhouY, Siliciano RF (2006) Nuclearretention of multiply spliced HIV-1 RNA in resting CD4+ T cells.PLoS Pathog 2:e68. doi:10.1371/journal.ppat.0020068

Lassen KG, Hebbeler AM, Bhattacharyya D, Lobritz MA, Greene WC(2012) A flexible model of HIV-1 latency permitting evaluation ofmany primary CD4 T-cell reservoirs. PLoS One 7:e30176.doi:10.1371/journal.pone.0030176

Lavender KJ et al (2013) BLT-humanized C57BL/6 Rag2−/−gammac−/−CD47−/− mice are resistant to GVHD and develop B- and T-cell immunity to HIV infection. Blood 122:4013–4020.doi:10.1182/blood-2013-06-506949

Lenasi T, Peterlin BM, Barboric M (2011) Cap-binding protein complexlinks pre-mRNA capping to transcription elongation and alternativesplicing through positive transcription elongation factor b (P-TEFb.J Biol Chem 286:22758–22768. doi:10.1074/jbc.M111.235077

Levere RD, Gong YF, Kappas A, Bucher DJ, Wormser GP, AbrahamNG(1991) Heme inhibits human immunodeficiency virus 1 replicationin cell cultures and enhances the antiviral effect of zidovudine. ProcNatl Acad Sci U S A 88:1756–1759

Lewinski MK et al (2005) Genome-wide analysis of chromosomal fea-tures repressing human immunodeficiency virus transcription. JVirol 79:6610–6619. doi:10.1128/JVI.79.11.6610-6619.2005

Lewinski MK et al (2006) Retroviral DNA integration: viral and cellulardeterminants of target-site selection. PLoS Pathog 2:e60.doi:10.1371/journal.ppat.0020060

Lewis MG et al (2011) Gold drug auranofin restricts the viral reservoir inthe monkey AIDS model and induces containment of viral loadfollowing ART suspension. AIDS 25:1347–1356. doi:10.1097/QAD.0b013e328347bd77

Lorenzo-Redondo R et al (2016) Persistent HIV-1 replication maintainsthe tissue reservoir during therapy. Nature 530:51–56. doi:10.1038/nature16933

LusicM,Marini B, Ali H, Lucic B, Luzzati R, GiaccaM (2013) Proximityto PML nuclear bodies regulates HIV-1 latency in CD4+ Tcells. CellHost Microbe 13:665–677. doi:10.1016/j.chom.2013.05.006

Maldarelli F et al (2007) ART suppresses plasma HIV-1 RNA to a stableset point predicted by pretherapy viremia. PLoS Pathog 3:e46.doi:10.1371/journal.ppat.0030046

Malhotra D, Fletcher AL, Turley SJ (2013) Stromal and hematopoieticcells in secondary lymphoid organs: partners in immunity. ImmunolRev 251:160–176. doi:10.1111/imr.12023

Marban C et al (2007) Recruitment of chromatin-modifying enzymes byCTIP2 promotes HIV-1 transcriptional silencing. EMBO J 26:412–423. doi:10.1038/sj.emboj.7601516

Margolis DM, Salzwedel K, Chomont N, Psomas C, Routy JP, Poli G,Lafeuillade A (2016) Highlights from the seventh internationalworkshop on HIV persistence during therapy, 8–11 December2015, Miami, Florida, USA. Journal of Virus Eradication 2:57–65

Maricato JT et al (2015) Epigenetic modulations in activated cells earlyafter HIV-1 infection and their possible functional consequences.PLoS One 10:e0119234. doi:10.1371/journal.pone.0119234

Marsden MD et al (2012) HIV latency in the humanized BLT mouse. JVirol 86:339–347. doi:10.1128/JVI.06366-11

Marsden MD et al (2015) Evaluation of HIV latency reversal using de-signed PKC modulators in humanized BLT mice. Journal of VirusEradication 1:1

Massanella M, Richman DD (2016) Measuring the latent reservoirin vivo. J Clin Invest 126:464–472. doi:10.1172/JCI80567

Massanella M, Fromentin R, Chomont N (2016) Residual inflammationand viral reservoirs: alliance against an HIV cure current opinion inHIVand. AIDS 11:234–241. doi:10.1097/COH.0000000000000230

Matsuda Y, Kobayashi-Ishihara M, Fujikawa D, Ishida T, Watanabe T,Yamagishi M (2015) Epigenetic heterogeneity in HIV-1 latency es-tablishment. Scientific Reports 5:7701. doi:10.1038/srep07701

Mbonye U, Karn J (2014) Transcriptional control of HIV latency: cellularsignaling pathways, epigenetics, happenstance and the hope for acure. Virology 454–455:328–339. doi:10.1016/j.virol.2014.02.008

McKinstry KK, Strutt TM, Swain SL (2010) The potential of CD4 T-cellmemory. Immunology 130:1–9. doi:10.1111/j.1365-2567.2010.03259.x

Mehla R et al (2010) Bryostatin modulates latent HIV-1 infection viaPKC and AMPK signaling but inhibits acute infection in a receptorindependent manner. PLoS One 5:e11160. doi:10.1371/journal.pone.0011160

Melkova Z, Bruckova M, Pitha PM (2000) The effect of nitric oxide onHIV-1 replication. Poster. First International Conference on Biology,Chemistry, and Therapeutic Applications of Nitric Oxide, SanFrancisco, USA

Melkova Z, Shankaran P, Hajkova V, Jilich D (2015a) Reactivation oflatent HIV-1 by iron-mediated redox stressh. Cytokine 76:92.doi:10.1016/j.cyto.2015.08.166

Melkova Z, Shankaran P, Hajkova V, Jilich D, Fujikura Y (2015b) Effectsof heme degradation products on reactivation of latent HIV-1.Journal of Virus Eradication 1 (Suppl. 1):42

Mendez-Lagares G, Jaramillo-Ruiz D, Pion M, Leal M, Munoz-Fernandez MA, Pacheco YM, Correa-Rocha R (2014) HIV

Folia Microbiol

Page 14: Current views on HIV-1 latency, persistence, and cure · et al. 2009; Fukazawa et al. 2015; Kim and Siliciano 2016; Van Lint et al. 2013).Additionally,thelatentreservoirisread-ily

infection deregulates the balance between regulatory T cells and IL-2-producing CD4 T cells by decreasing the expression of the IL-2receptor in Treg. J Acquir Immune Defic Syndr 65:278–282.doi:10.1097/QAI.0000000000000092

Metcalf Pate KA et al (2015) A murine viral outgrowth assay to detectresidual HIV type 1 in patients with undetectable viral loads. TheJournal of Infectious Diseases 212:1387–1396. doi:10.1093/infdis/jiv230

Miles B et al (2015) Follicular regulatory Tcells impair follicular T helpercells in HIV and SIV infection. Nat Commun 6:8608. doi:10.1038/ncomms9608

Modjarrad K, Vermund SH (2010) Effect of treating co-infections onHIV-1 viral load: a systematic review. Lancet Infect Dis 10:455–463. doi:10.1016/S1473-3099(10)70093-1

MonroeKM, Yang Z, Johnson JR, GengX,Doitsh G, KroganNJ, GreeneWC (2014) IFI16 DNA sensor is required for death of lymphoidCD4 T cells abortively infected with HIV. Science 343:428–432.doi:10.1126/science.1243640

Mottamal M, Zheng S, Huang TL, Wang G (2015) Histone deacetylaseinhibitors in clinical studies as templates for new anticancer agents.Molecules 20:3898–3941. doi:10.3390/molecules20033898

Mousseau G, Kessing CF, Fromentin R, Trautmann L, Chomont N,Valente ST (2015) The Tat inhibitor didehydro-cortistatin A pre-vents HIV-1 reactivation from latency. MBio 6:e00465.doi:10.1128/mBio.00465-15

Murray JM et al (2014) HIV DNA subspecies persist in both activatedand resting memory CD4+ T cells during antiretroviral therapy. JVirol 88:3516–3526. doi:10.1128/JVI.03331-13

Ndung’u Tet al (2001) Infectious simian/human immunodeficiency viruswith human immunodeficiency virus type 1 subtype C from anAfrican isolate: rhesus macaque model. J Virol 75:11417–11425.doi:10.1128/JVI.75.23.11417-11425.2001

Omoto S et al (2004) HIV-1 nef suppression by virally encodedmicroRNA. Retrovirology 1:44. doi:10.1186/1742-4690-1-44

Ott M, Verdin E (2013) Three rules for HIV latency: location, location,and location. Cell Host Microbe 13:625–626. doi:10.1016/j.chom.2013.05.016

Pace GW, Leaf CD (1995) The role of oxidative stress in HIV disease.Free Radic Biol Med 19:523–528

Palmer S et al (2003) New real-time reverse transcriptase-initiated PCRassay with single-copy sensitivity for human immunodeficiency vi-rus type 1 RNA in plasma. J Clin Microbiol 41:4531–4536

Palmer S et al (2008) Low-level viremia persists for at least 7 years inpatients on suppressive antiretroviral therapy. Proc Natl Acad Sci US A 105:3879–3884. doi:10.1073/pnas.0800050105

Pasternak AO, Adema KW, Bakker M, Jurriaans S, Berkhout B,Cornelissen M, Lukashov VV (2008) Highly sensitive methodsbased on seminested real-time reverse transcription-PCR for quan-titation of human immunodeficiency virus type 1 unspliced andmultiply spliced RNA and proviral DNA. J Clin Microbiol 46:2206–2211. doi:10.1128/JCM.00055-08

Pasternak AO, Lukashov VV, Berkhout B (2013) Cell-associated HIVRNA: a dynamic biomarker of viral persistence. Retrovirology 10:41. doi:10.1186/1742-4690-10-41

Perelson AS, Neumann AU, Markowitz M, Leonard JM, Ho DD (1996)HIV-1 dynamics in vivo: virion clearance rate, infected cell life-span, and viral generation time. Science 271:1582–1586

Perelson AS et al (1997) Decay characteristics of HIV-1-infected com-partments during combination therapy. Nature 387:188–191.doi:10.1038/387188a0

Perkins ND, Edwards NL, Duckett CS, Agranoff AB, Schmid RM, NabelGJ (1993) A cooperative interaction between NF-kappa B and Sp1is required for HIV-1 enhancer activation. EMBO J 12:3551–3558

Ploquin MJ, Silvestri G, Muller-Trutwin M (2016) Immune activation inHIV infect ion: what can the natural hosts of simian

immunodeficiency virus teach us? Current opinion in HIV and.AIDS 11:201–208. doi:10.1097/COH.0000000000000238

Policicchio BB, Pandrea I, Apetrei C (2016) Animal models for HIV cureresearch. Front Immunol 7:12. doi:10.3389/fimmu.2016.00012

Price DH (2000) P-TEFb, a cyclin-dependent kinase controlling elonga-tion by RNA polymerase II. Mol Cell Biol 20:2629–2634

Procopio FA et al (2015) A novel assay to measure the magnitude of theinducible viral reservoir in HIV-infected individuals. EBioMedicine2:872–881. doi:10.1016/j.ebiom.2015.06.019

Pyo CW, Yang YL, Yoo NK, Choi SY (2008) Reactive oxygen speciesactivate HIV long terminal repeat via post-translational control ofNF-kappaB. Biochem Biophys Res Commun 376:180–185.doi:10.1016/j.bbrc.2008.08.114

Rasmussen TA et al (2013) Comparison of HDAC inhibitors in clinicaldevelopment: effect on HIV production in latently infected cells andT-cell activation. Human Vaccines & Immunotherapeutics 9:993–1001. doi:10.4161/hv.23800

Rasmussen TA et al (2014) Panobinostat, a histone deacetylase inhibitor,for latent-virus reactivation in HIV-infected patients on suppressiveantiretroviral therapy: a phase 1/2, single group, clinical trial. TheLancet HIV 1:e13–e21. doi:10.1016/S2352-3018(14)70014-1

Romerio F, Zapata JC (2015) Detection and enrichment to near purity ofrare HIV-1 infected cells by PrimeFlow RNA. Journal of VirusEradication 1:5

Saison J et al (2014) Association between discordant immunologicalresponse to highly active anti-retroviral therapy, regulatory T cellpercentage, immune cell activation and very low-level viraemia inHIV-infected patients. Clin Exp Immunol 176:401–409.doi:10.1111/cei.12278

Saleh S, Solomon A, Wightman F, Xhilaga M, Cameron PU, Lewin SR(2007) CCR7 ligands CCL19 and CCL21 increase permissivenessof resting memory CD4+ T cells to HIV-1 infection: a novel modelof HIV-1 latency. Blood 110:4161–4164. doi:10.1182/blood-2007-06-097907

Sanchez G, Xu X, Chermann JC, Hirsch I (1997) Accumulation of de-fective viral genomes in peripheral blood mononuclear cells of hu-man immunodeficiency virus type 1-infected individuals. J Virol 71:2233–2240

Schopman NC et al (2012) Deep sequencing of virus-infected cells re-veals HIV-encoded small RNAs. Nucleic Acids Res 40:414–427.doi:10.1093/nar/gkr719

Schuetz A et al (2014) Initiation of ART during early acute HIV infectionpreserves mucosal Th17 function and reverses HIV-related immuneactivation. PLoS Pathog 10:e1004543. doi:10.1371/journal.ppat.1004543

Serrao E, Engelman AN (2016) Sites of retroviral DNA integration: frombasic research to clinical applications. Crit Rev Biochem Mol Biol51:26–42. doi:10.3109/10409238.2015.1102859

Shan L, Siliciano RF (2013) From reactivation of latent HIV-1 to elimi-nation of the latent reservoir: the presence of multiple barriers toviral eradication. BioEssays : News and Reviews in Molecular,Cellular and Developmental Biology 35:544–552. doi:10.1002/bies.201200170

Shan L et al (2011) Influence of host gene transcription level and orien-tation on HIV-1 latency in a primary-cell model. J Virol 85:5384–5393. doi:10.1128/JVI.02536-10

Shan L, Rabi SA, Laird GM, Eisele EE, Zhang H, Margolick JB,Siliciano RF (2013) A novel PCR assay for quantification of HIV-1 RNA. J Virol 87:6521–6525. doi:10.1128/JVI.00006-13

Shankaran P, Vlkova L, Liskova J, Melkova Z (2011) Heme arginatepotentiates latent HIV-1 reactivation while inhibiting the acute infec-tion. Antivir Res 92:434–446. doi:10.1016/j.antiviral.2011.09.011

Sharp PM, Hahn BH (2011) Origins of HIV and the AIDS pandemic.Cold Spring Harbor Perspectives in Medicine 1:a006841.doi:10.1101/cshperspect.a006841

Folia Microbiol

Page 15: Current views on HIV-1 latency, persistence, and cure · et al. 2009; Fukazawa et al. 2015; Kim and Siliciano 2016; Van Lint et al. 2013).Additionally,thelatentreservoirisread-ily

Siliciano JD, Siliciano RF (2005) Enhanced culture assay for detectionand quantitation of latently infected, resting CD4+ T-cells carryingreplication-competent virus in HIV-1-infected individuals. MethodsMol Biol 304:3–15. doi:10.1385/1-59259-907-9:003

Siliciano JD et al (2003) Long-term follow-up studies confirm the stabil-ity of the latent reservoir for HIV-1 in resting CD4+ Tcells. Nat Med9:727–728. doi:10.1038/nm880

Sloan DD et al (2015) Targeting HIV reservoir in infected CD4 Tcells bydual-affinity re-targeting molecules (DARTs) that bind HIV enve-lope and recruit cytotoxic T cells. PLoS Pathog 11:e1005233.doi:10.1371/journal.ppat.1005233

Sogaard OS et al (2015) The depsipeptide romidepsin reverses HIV-1latency in vivo. PLoS Pathog 11:e1005142. doi:10.1371/journal.ppat.1005142

Soriano-Sarabia N et al (2015) Peripheral Vgamma9Vdelta2 T cells are anovel reservoir of latent HIV infection. PLoS Pathog 11:e1005201.doi:10.1371/journal.ppat.1005201

Spina CA et al (2013) An in-depth comparison of latent HIV-1 reactiva-tion in multiple cell model systems and resting CD4+ T cells fromaviremic patients. PLoS Pathog 9:e1003834. doi:10.1371/journal.ppat.1003834

Spivak AM et al (2014) A pilot study assessing the safety and latency-reversing activity of disulfiram in HIV-1-infected adults on antire-troviral therapy. Clinical Infectious Diseases : an OfficialPublication of the Infectious Diseases Society of America 58:883–890. doi:10.1093/cid/cit813

Sung TL, Rice AP (2009) miR-198 inhibits HIV-1 gene expression andreplication in monocytes and its mechanism of action appears toinvolve repression of cyclin T1. PLoS Pathog 5:e1000263.doi:10.1371/journal.ppat.1000263

Tae S, Karkhanis V, Velasco K, Yaneva M, Erdjument-Bromage H,Tempst P, Sif S (2011) Bromodomain protein 7 interacts withPRMT5 and PRC2, and is involved in transcriptional repression oftheir target genes. Nucleic Acids Res 39:5424–5438. doi:10.1093/nar/gkr170

Thompson MR et al (2014) Interferon gamma-inducible protein (IFI) 16transcriptionally regulates type i interferons and other interferon-stimulated genes and controls the interferon response to both DNAand RNAviruses. J Biol Chem 289:23568–23581. doi:10.1074/jbc.M114.554147

Traggiai E, Chicha L, Mazzucchelli L, Bronz L, Piffaretti JC,Lanzavecchia A, Manz MG (2004) Development of a human adap-tive immune system in cord blood cell-transplanted mice. Science304:104–107. doi:10.1126/science.1093933

Tran TA et al (2008) Resting regulatory CD4 T cells: a site of HIVpersistence in patients on long-term effective antiretroviral therapy.PLoS One 3:e3305. doi:10.1371/journal.pone.0003305

Trejbalova K et al (2016) Development of 5′ LTR DNA methylationof latent HIV-1 provirus in cell line models and in long-term-infected individuals. Clinical Epigenetics 8. doi:10.1186/s13148-016-0185-6

Triboulet R et al (2007) Suppression of microRNA-silencing pathway byHIV-1 during virus replication. Science 315:1579–1582.doi:10.1126/science.1136319

Tyagi M, Karn J (2007) CBF-1 promotes transcriptional silencing duringthe establishment of HIV-1 latency. EMBO J 26:4985–4995.doi:10.1038/sj.emboj.7601928

Tyagi M, Pearson RJ, Karn J (2010) Establishment of HIV latency inprimary CD4+ cells is due to epigenetic transcriptional silencingand P-TEFb restriction. J Virol 84:6425–6437. doi:10.1128/JVI.01519-09

Van Lint C, Bouchat S, Marcello A (2013) HIV-1 transcription and laten-cy: an update. Retrovirology 10:67. doi:10.1186/1742-4690-10-67

Verdin E, Paras P Jr, Van Lint C (1993) Chromatin disruption in thepromoter of human immunodeficiency virus type 1 during transcrip-tional activation. EMBO J 12:3249–3259

Vire E et al (2006) The polycomb group protein EZH2 directlycontrols DNA methylation. Nature 439:871–874. doi:10.1038/nature04431

Wang L, de Zoeten EF, Greene MI, Hancock WW (2009)Immunomodulatory effects of deacetylase inhibitors: therapeutictargeting of FOXP3+ regulatory T cells. Nat Rev Drug Discov 8:969–981. doi:10.1038/nrd3031

Wang X, Cho B, Suzuki K, Xu Y, Green JA, An J, Cyster JG (2011)Follicular dendritic cells help establish follicle identity and promoteB cell retention in germinal centers. J Exp Med 208:2497–2510.doi:10.1084/jem.20111449

Wei X et al (1995) Viral dynamics in human immunodeficiency virustype 1 infection. Nature 373:117–122. doi:10.1038/373117a0

Wei P, Garber ME, Fang SM, Fischer WH, Jones KA (1998) A novelCDK9-associated C-type cyclin interacts directly with HIV-1 tat andmediates its high-affinity, loop-specific binding to TAR RNA. Cell92:451–462

Weinberger LS (2015) A minimal fate-selection switch. Curr Opin CellBiol 37:111–118. doi:10.1016/j.ceb.2015.10.005

Weinberger LS, Burnett JC, Toettcher JE, Arkin AP, Schaffer DV (2005)Stochastic gene expression in a lentiviral positive-feedback loop:HIV-1 Tat fluctuations drive phenotypic diversity. Cell 122:169–182. doi:10.1016/j.cell.2005.06.006

Weinberger LS, Dar RD, Simpson ML (2008) Transient-mediated fatedetermination in a transcriptional circuit of HIV. Nat Genet 40:466–470. doi:10.1038/ng.116

Wightman F et al (2010) Both CD31(+) and CD31(−) naive CD4(+) Tcells are persistent HIV type 1-infected reservoirs in individualsreceiving antiretroviral therapy. The Journal of Infectious Diseases202:1738–1748. doi:10.1086/656721

Williams SA, Chen LF, Kwon H, Ruiz-Jarabo CM, Verdin E, GreeneWC(2006) NF-kappaB p50 promotes HIV latency through HDAC re-cruitment and repression of transcriptional initiation. EMBO J 25:139–149. doi:10.1038/sj.emboj.7600900

Wires ES, Alvarez D, Dobrowolski C, Wang Y, Morales M, Karn J,Harvey BK (2012) Methamphetamine activates nuclear factorkappa-light-chain-enhancer of activated B cells (NF-kappaB) andinduces human immunodeficiency virus (HIV) transcription in hu-man microglial cells. Journal of Neurovirology 18:400–410.doi:10.1007/s13365-012-0103-4

Xing S et al (2011) Disulfiram reactivates latent HIV-1 in a Bcl-2-transduced primary CD4+ T cell model without inducingglobal T cell activation. J Virol 85:6060–6064. doi:10.1128/JVI.02033-10

Yamada T, Yamaguchi Y, Inukai N, Okamoto S,Mura T, Handa H (2006)P-TEFb-mediated phosphorylation of hSpt5 C-terminal repeats iscritical for processive transcription elongation. Mol Cell 21:227–237. doi:10.1016/j.molcel.2005.11.024

Yang HC, Shen L, Siliciano RF, Pomerantz JL (2009a) Isolation of acellular factor that can reactivate latent HIV-1 without T cell activa-tion. Proc Natl Acad Sci U S A 106:6321–6326. doi:10.1073/pnas.0809536106

Yang HC et al (2009b) Small-molecule screening using a human primarycell model of HIV latency identifies compounds that reverse latencywithout cellular activation. J Clin Invest 119:3473–3486.doi:10.1172/JCI39199

Yoh SM et al (2015) PQBP1 is a proximal sensor of the cGAS-dependentinnate response to HIV-1. Cell 161:1293–1305. doi:10.1016/j.cell.2015.04.050

Zhang J, Scadden DT, Crumpacker CS (2007) Primitive hematopoieticcells resist HIV-1 infection via p21. J Clin Invest 117:473–481.doi:10.1172/JCI28971

Zolotukhin AS et al (2003) PSF acts through the human immunodeficien-cy virus type 1 mRNA instability elements to regulate virus expres-sion. Mol Cell Biol 23:6618–6630

Folia Microbiol